Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
BMC Cancer ; 23(1): 775, 2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37596538

ABSTRACT

BACKGROUND: Inhibitors of Poly (ADP-Ribose) Polymerases (PARP) provide clinical benefit to patients with breast and ovarian cancers, by compromising the DNA repair activity of cancer cells. Although these agents extend progression-free survival in many patients, responses can be short lived with many patients ultimately progressing. Identification of combination partners that increase dependence of cancer cells to the DNA repair activity of PARPs may represent a strategy to increase the utility of PARP inhibitors. Protein arginine methyltransferase 5 (PRMT5) regulates DNA damage response pathways through splicing and protein modification, and inhibitors of PRMT5 have recently entered clinical trials. METHODS: The effect of PRMT5 inhibition on the levels of DNA damage and repair markers including γH2AX, RAD51, and 53BP1 was determined using high content immunofluorescent imaging. The anti-proliferative activity of the combination of PRMT5 and PARP inhibitors was evaluated using in vitro models of breast and ovarian cancers using both cell lines and ex vivo patient derived xenografts. Finally, the combinations of PRMT5 and PARP inhibitors were evaluated in cell line xenograft models in vivo. RESULTS: Inhibition of PRMT5 by GSK3326595 led to increased levels of markers of DNA damage. The addition of GSK3326595 to the PARP inhibitor, niraparib, resulted in increased growth inhibition of breast and ovarian cancer cell lines and patient derived spheroids. In vivo, the combination improved the partial effects on tumor growth inhibition achieved by either single agent, producing complete tumor stasis and regression. CONCLUSION: These data demonstrate that inhibition of PRMT5 induced signatures of DNA damage in models of breast and ovarian cancer. Furthermore, combination with the PARP inhibitor, Niraparib, resulted in increased anti-tumor activity in vitro and in vivo. Overall, these data suggest inhibition of PRMT5 as a mechanism to broaden and enhance the clinical application of PARP inhibitors.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Humans , Female , Animals , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Ovarian Neoplasms/drug therapy , DNA Damage , Disease Models, Animal , Protein-Arginine N-Methyltransferases
2.
J Biol Chem ; 297(2): 100928, 2021 08.
Article in English | MEDLINE | ID: mdl-34274316

ABSTRACT

B-cell lymphoma 6 (BCL6) is a zinc finger transcriptional repressor possessing a BTB-POZ (BR-C, ttk, and bab for BTB; pox virus and zinc finger for POZ) domain, which is required for homodimerization and association with corepressors. BCL6 has multiple roles in normal immunity, autoimmunity, and some types of lymphoma. Mice bearing disrupted BCL6 loci demonstrate suppressed high-affinity antibody responses to T-dependent antigens. The corepressor binding groove in the BTB-POZ domain is a potential target for small compound-mediated therapy. Several inhibitors targeting this binding groove have been described, but these compounds have limited or absent in vivo activity. Biophysical studies of a novel compound, GSK137, showed an in vitro pIC50 of 8 and a cellular pIC50 of 7.3 for blocking binding of a peptide derived from the corepressor silencing mediator for retinoid or thyroid hormone receptors to the BCL6 BTB-POZ domain. The compound has good solubility (128 µg/ml) and permeability (86 nM/s). GSK137 caused little change in cell viability or proliferation in four BCL6-expressing B-cell lymphoma lines, although there was modest dose-dependent accumulation of G1 phase cells. Pharmacokinetic studies in mice showed a profile compatible with achieving good levels of target engagement. GSK137, administered orally, suppressed immunoglobulin G responses and reduced numbers of germinal centers and germinal center B cells following immunization of mice with the hapten trinitrophenol. Overall, we report a novel small-molecule BCL6 inhibitor with in vivo activity that inhibits the T-dependent antigen immune response.


Subject(s)
Proto-Oncogene Proteins c-bcl-6 , Animals , B-Lymphocytes/metabolism , Humans , Mice , Transcription, Genetic , Zinc Fingers
3.
Int J Cancer ; 150(6): 993-1006, 2022 03 15.
Article in English | MEDLINE | ID: mdl-34724226

ABSTRACT

Molibresib is an orally bioavailable, selective, small molecule BET protein inhibitor. Results from a first time in human study in solid tumors resulted in the selection of a 75 mg once daily dose of the besylate formulation of molibresib as the recommended Phase 2 dose (RP2D). Here we present the results of Part 2 of our study, investigating safety, pharmacokinetics, pharmacodynamics and clinical activity of molibresib at the RP2D for nuclear protein in testis carcinoma (NC), small cell lung cancer, castration-resistant prostate cancer (CRPC), triple-negative breast cancer, estrogen receptor-positive breast cancer and gastrointestinal stromal tumor. The primary safety endpoints were incidence of adverse events (AEs) and serious AEs; the primary efficacy endpoint was overall response rate. Secondary endpoints included plasma concentrations and gene set enrichment analysis (GSEA). Molibresib 75 mg once daily demonstrated no unexpected toxicities. The most common treatment-related AEs (any grade) were thrombocytopenia (64%), nausea (43%) and decreased appetite (37%); 83% of patients required dose interruptions and 29% required dose reductions due to AEs. Antitumor activity was observed in NC and CRPC (one confirmed partial response each, with observed reductions in tumor size), although predefined clinically meaningful response rates were not met for any tumor type. Total active moiety median plasma concentrations after single and repeated administration were similar across tumor cohorts. GSEA revealed that gene expression changes with molibresib varied by patient, response status and tumor type. Investigations into combinatorial approaches that use BET inhibition to eliminate resistance to other targeted therapies are warranted.


Subject(s)
Benzodiazepines/therapeutic use , Neoplasms/drug therapy , Nuclear Proteins/metabolism , Testicular Neoplasms/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Benzodiazepines/administration & dosage , Benzodiazepines/adverse effects , Benzodiazepines/pharmacokinetics , Female , Humans , Male , Middle Aged , Nerve Tissue Proteins/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Young Adult
4.
Pediatr Blood Cancer ; 67(6): e28267, 2020 06.
Article in English | MEDLINE | ID: mdl-32307821

ABSTRACT

BACKGROUND: The treatment of high-risk neuroblastoma continues to present a formidable challenge to pediatric oncology. Previous studies have shown that Bromodomain and extraterminal (BET) inhibitors can inhibit MYCN expression and suppress MYCN-amplified neuroblastoma in vivo. Furthermore, alterations within RAS-MAPK (mitogen-activated protein kinase) signaling play significant roles in neuroblastoma initiation, maintenance, and relapse, and mitogen-activated extracellular signal-regulated kinase (MEK) inhibitors demonstrate efficacy in subsets of neuroblastoma preclinical models. Finally, hyperactivation of RAS-MAPK signaling has been shown to promote resistance to BET inhibitors. Therefore, we examined the antitumor efficacy of combined BET/MEK inhibition utilizing I-BET726 or I-BET762 and trametinib in high-risk neuroblastoma. PROCEDURE: Utilizing a panel of genomically annotated neuroblastoma cell line models, we investigated the in vitro effects of combined BET/MEK inhibition on cell proliferation and apoptosis. Furthermore, we evaluated the effects of combined inhibition in neuroblastoma xenograft models. RESULTS: Combined BET and MEK inhibition demonstrated synergistic effects on the growth and survival of a large panel of neuroblastoma cell lines through augmentation of apoptosis. A combination therapy slowed tumor growth in a non-MYCN-amplified, NRAS-mutated neuroblastoma xenograft model, but had no efficacy in an MYCN-amplified model harboring a loss-of-function mutation in NF1. CONCLUSIONS: Combinatorial BET and MEK inhibition was synergistic in the vast majority of neuroblastoma cell lines in the in vitro setting but showed limited antitumor activity in vivo. Collectively, these data do not support clinical development of this combination in high-risk neuroblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Benzodiazepines/pharmacology , MAP Kinase Kinase 1/antagonists & inhibitors , Neuroblastoma/drug therapy , Proteins/antagonists & inhibitors , Pyridones/pharmacology , Pyrimidinones/pharmacology , Animals , Apoptosis , Cell Proliferation , Female , Humans , Mice , Mice, SCID , Neuroblastoma/metabolism , Neuroblastoma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 30(2): 334-343, 2024 01 17.
Article in English | MEDLINE | ID: mdl-37992310

ABSTRACT

PURPOSE: Endocrine-based therapy is the initial primary treatment option for hormone receptor-positive and human epidermal growth factor receptor 2-negative (HR+/HER2-) metastatic breast cancer (mBC). However, patients eventually experience disease progression due to resistance to endocrine therapy. Molibresib (GSK525762) is a small-molecule inhibitor of bromodomain and extraterminal (BET) family proteins (BRD2, BRD3, BRD4, and BRDT). Preclinical data suggested that the combination of molibresib with endocrine therapy might overcome endocrine resistance. This study aimed to investigate the safety, tolerability, pharmacokinetics, pharmacodynamics, and efficacy [objective response rate (ORR)] of molibresib combined with fulvestrant in women with HR+/HER2- mBC. PATIENTS AND METHODS: In this phase I/II dose-escalation and dose-expansion study, patients received oral molibresib 60 or 80 mg once daily in combination with intramuscular fulvestrant. Patients enrolled had relapsed/refractory, advanced/metastatic HR+/HER2- breast cancer with disease progression on prior treatment with an aromatase inhibitor, with or without a cyclin-dependent kinase 4/6 inhibitor. RESULTS: The study included 123 patients. The most common treatment-related adverse events (AE) were nausea (52%), dysgeusia (49%), and fatigue (45%). At a 60-mg dosage of molibresib, >90% of patients experienced treatment-related AE. Grade 3 or 4 treatment-related AE were observed in 47% and 48% of patients treated with molibresib 60 mg and molibresib 80 mg, respectively. The ORR was 13% [95% confidence interval (CI), 8-20], not meeting the 25% threshold for proceeding to phase II. Among 82 patients with detected circulating tumor DNA and clinical outcome at study enrollment, a strong association was observed between the detection of copy-number amplification and poor progression-free survival (HR, 2.89; 95% CI, 1.73-4.83; P < 0.0001). CONCLUSIONS: Molibresib in combination with fulvestrant did not demonstrate clinically meaningful activity in this study.


Subject(s)
Benzodiazepines , Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Fulvestrant , Nuclear Proteins , Receptor, ErbB-2/metabolism , Transcription Factors , Disease Progression , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bromodomain Containing Proteins , Cell Cycle Proteins
6.
Clin Cancer Res ; 29(4): 711-722, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36350312

ABSTRACT

PURPOSE: Molibresib is a selective, small molecule inhibitor of the bromodomain and extra-terminal (BET) protein family. This was an open-label, two-part, Phase I/II study investigating molibresib monotherapy for the treatment of hematological malignancies (NCT01943851). PATIENTS AND METHODS: Part 1 (dose escalation) determined the recommended Phase 2 dose (RP2D) of molibresib in patients with acute myeloid leukemia (AML), Non-Hodgkin lymphoma (NHL), or multiple myeloma. Part 2 (dose expansion) investigated the safety and efficacy of molibresib at the RP2D in patients with relapsed/refractory myelodysplastic syndrome (MDS; as well as AML evolved from antecedent MDS) or cutaneous T-cell lymphoma (CTCL). The primary endpoint in Part 1 was safety and the primary endpoint in Part 2 was objective response rate (ORR). RESULTS: There were 111 patients enrolled (87 in Part 1, 24 in Part 2). Molibresib RP2Ds of 75 mg daily (for MDS) and 60 mg daily (for CTCL) were selected. Most common Grade 3+ adverse events included thrombocytopenia (37%), anemia (15%), and febrile neutropenia (15%). Six patients achieved complete responses [3 in Part 1 (2 AML, 1 NHL), 3 in Part 2 (MDS)], and 7 patients achieved partial responses [6 in Part 1 (4 AML, 2 NHL), 1 in Part 2 (MDS)]. The ORRs for Part 1, Part 2, and the total study population were 10% [95% confidence interval (CI), 4.8-18.7], 25% (95% CI, 7.3-52.4), and 13% (95% CI, 6.9-20.6), respectively. CONCLUSIONS: While antitumor activity was observed with molibresib, use was limited by gastrointestinal and thrombocytopenia toxicities. Investigations of molibresib as part of combination regimens may be warranted.


Subject(s)
Hematologic Neoplasms , Leukemia, Myeloid, Acute , Lymphoma, Non-Hodgkin , Thrombocytopenia , Humans , Lymphoma, Non-Hodgkin/drug therapy , Hematologic Neoplasms/drug therapy , Leukemia, Myeloid, Acute/drug therapy
7.
J Med Chem ; 64(15): 10772-10805, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34255512

ABSTRACT

The profound efficacy of pan-BET inhibitors is well documented, but these epigenetic agents have shown pharmacology-driven toxicity in oncology clinical trials. The opportunity to identify inhibitors with an improved safety profile by selective targeting of a subset of the eight bromodomains of the BET family has triggered extensive medicinal chemistry efforts. In this article, we disclose the identification of potent and selective drug-like pan-BD2 inhibitors such as pyrazole 23 (GSK809) and furan 24 (GSK743) that were derived from the pyrrole fragment 6. We transpose the key learnings from a previous pyridone series (GSK620 2 as a representative example) to this novel class of inhibitors, which are characterized by significantly improved solubility relative to our previous research.


Subject(s)
Furans/pharmacology , Proteins/antagonists & inhibitors , Pyrazoles/pharmacology , Dose-Response Relationship, Drug , Furans/chemistry , Humans , Molecular Structure , Proteins/metabolism , Pyrazoles/chemistry , Structure-Activity Relationship
8.
JNCI Cancer Spectr ; 4(2): pkz093, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32328561

ABSTRACT

BACKGROUND: Bromodomain and extra-terminal domain proteins are promising epigenetic anticancer drug targets. This first-in-human study evaluated the safety, recommended phase II dose, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of the bromodomain and extra-terminal domain inhibitor molibresib (GSK525762) in patients with nuclear protein in testis (NUT) carcinoma (NC) and other solid tumors. METHODS: This was a phase I and II, open-label, dose-escalation study. Molibresib was administered orally once daily. Single-patient dose escalation (from 2 mg/d) was conducted until the first instance of grade 2 or higher drug-related toxicity, followed by a 3 + 3 design. Pharmacokinetic parameters were obtained during weeks 1 and 3. Circulating monocyte chemoattractant protein-1 levels were measured as a pharmacodynamic biomarker. RESULTS: Sixty-five patients received molibresib. During dose escalation, 11% experienced dose-limiting toxicities, including six instances of grade 4 thrombocytopenia, all with molibresib 60-100 mg. The most frequent treatment-related adverse events of any grade were thrombocytopenia (51%) and gastrointestinal events, including nausea, vomiting, diarrhea, decreased appetite, and dysgeusia (22%-42%), anemia (22%), and fatigue (20%). Molibresib demonstrated an acceptable safety profile up to 100 mg; 80 mg once daily was selected as the recommended phase II dose. Following single and repeat dosing, molibresib showed rapid absorption and elimination (maximum plasma concentration: 2 hours; t1/2: 3-7 hours). Dose-dependent reductions in circulating monocyte chemoattractant protein-1 levels were observed. Among 19 patients with NC, four achieved either confirmed or unconfirmed partial response, eight had stable disease as best response, and four were progression-free for more than 6 months. CONCLUSIONS: Once-daily molibresib was tolerated at doses demonstrating target engagement. Preliminary data indicate proof-of-concept in NC.

9.
Mol Cell Biol ; 25(3): 1162-72, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15657441

ABSTRACT

The SAGA complex is a multisubunit protein complex involved in transcriptional regulation in Saccharomyces cerevisiae. SAGA combines proteins involved in interactions with DNA-bound activators and TATA-binding protein (TBP), as well as enzymes for histone acetylation (Gcn5) and histone deubiquitylation (Ubp8). We recently showed that H2B ubiquitylation and Ubp8-mediated deubiquitylation are both required for transcriptional activation. For this study, we investigated the interaction of Ubp8 with SAGA. Using mutagenesis, we identified a putative zinc (Zn) binding domain within Ubp8 as being critical for the association with SAGA. The Zn binding domain is required for H2B deubiquitylation and for growth on media requiring Ubp8's function in gene activation. Furthermore, we identified an 11-kDa subunit of SAGA, Sgf11, and showed that it is required for the Ubp8 association with SAGA and for H2B deubiquitylation. Different approaches indicated that the functions of Ubp8 and Sgf11 are related and separable from those of other components of SAGA. In particular, the profiles of Ubp8 and Sgf11 deletions were remarkably similar in microarray analyses and synthetic genetic interactions and were distinct from those of the Spt3 and Spt8 subunits of SAGA, which are involved in TBP regulation. These data indicate that Ubp8 and Sgf11 likely represent a new functional module within SAGA that is involved in gene regulation through H2B deubiquitylation.


Subject(s)
Histones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Fungal/physiology , Histone Acetyltransferases , Microarray Analysis , Molecular Sequence Data , Mutation/genetics , Protein Kinases/metabolism , Transcriptional Activation , Zinc/metabolism
10.
Structure ; 14(3): 477-85, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16531232

ABSTRACT

The Snf1/AMPK kinases are intracellular energy sensors, and the AMPK pathway has been implicated in a variety of metabolic human disorders. Here we report the crystal structure of the kinase domain from yeast Snf1, revealing a bilobe kinase fold with greatest homology to cyclin-dependant kinase-2. Unexpectedly, the crystal structure also reveals a novel homodimer that we show also forms in solution, as demonstrated by equilibrium sedimentation, and in yeast cells, as shown by coimmunoprecipitation of differentially tagged intact Snf1. A mapping of sequence conservation suggests that dimer formation is a conserved feature of the Snf1/AMPK kinases. The conformation of the conserved alphaC helix, and the burial of the activation segment and substrate binding site within the dimer, suggests that it represents an inactive form of the kinase. Taken together, these studies suggest another layer of kinase regulation within the Snf1/AMPK family, and an avenue for development of AMPK-specific activating compounds.


Subject(s)
Adenylate Kinase/chemistry , Cyclin-Dependent Kinase 2/chemistry , Yeasts/genetics , Adenosine Triphosphate/chemistry , Adenylate Kinase/genetics , Amino Acid Sequence , Crystallography , Dimerization , Molecular Sequence Data , Multienzyme Complexes/chemistry , Phosphotransferases/chemistry , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Solutions , Structural Homology, Protein , Structure-Activity Relationship , Yeasts/chemistry
11.
Oncogenesis ; 7(4): 35, 2018 Apr 20.
Article in English | MEDLINE | ID: mdl-29674704

ABSTRACT

BET inhibitors exhibit broad activity in cancer models, making predictive biomarkers challenging to define. Here we investigate the biomarkers of activity of the clinical BET inhibitor GSK525762 (I-BET; I-BET762) across cancer cell lines and demonstrate that KRAS mutations are novel resistance biomarkers. This finding led us to combine BET with RAS pathway inhibition using MEK inhibitors to overcome resistance, which resulted in synergistic effects on growth and survival in RAS pathway mutant models as well as a subset of cell lines lacking RAS pathway mutations. GSK525762 treatment up-regulated p-ERK1/2 levels in both RAS pathway wild-type and mutant cell lines, suggesting that MEK/ERK pathway activation may also be a mechanism of adaptive BET inhibitor resistance. Importantly, gene expression studies demonstrated that the BET/MEK combination uniquely sustains down-regulation of genes associated with mitosis, leading to prolonged growth arrest that is not observed with either single agent therapy. These studies highlight a potential to enhance the clinical benefit of BET and MEK inhibitors and provide a strong rationale for clinical evaluation of BET/MEK combination therapies in cancer.

12.
Novartis Found Symp ; 259: 63-73; discussion 73-7, 163-9, 2004.
Article in English | MEDLINE | ID: mdl-15171247

ABSTRACT

Previous models for the role of histone modifications suggest that adding and removing modifications, such as acetylation/deacetylation in gene regulation, are functionally antagonistic. We have investigated a transcriptional role of H2B C-terminal ubiquitylation and de-ubiquitylation in Saccharomyces cerevisiae. H2B ubiquitylation is required for optimal transcription of SUC2 and GAL1 genes. The ubiquitin hydrolase Ubp8 is a stable component of SAGA but not ADA complexes, and is not required for overall integrity of SAGA. Biochemical and genetic evidence indicates that Ubp8 targets H2B for deubiquitylation. The dynamic balance of H2B ubiquitylation/deubiquitylation is important for GAL1 transcription since either substitution of the ubiquitylation site in H2B (Lys123), or loss of Ubp8, lowers GAL1 expression. Further, this balance of ubiquitylation appears to set the balance of histone H3 methylation at Lys4 relative to Lys36. Thus, unlike acetylation/deacetylation whose functions are mutually opposing, both ubiquitylation and de-ubiquitylation are required for gene activation. These results suggest that ubiquitylation of histones has a unique role among histone modifications, possibly to orchestrate an ordered pathway of chromatin alterations.


Subject(s)
Gene Expression Regulation, Fungal/physiology , Histones/metabolism , Ubiquitin/metabolism , DNA Methylation , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/genetics , Transcriptional Activation , beta-Fructofuranosidase/biosynthesis , beta-Fructofuranosidase/genetics
13.
Oncotarget ; 4(12): 2419-29, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24293458

ABSTRACT

BET (bromodomain and extra-terminal) proteins regulate gene expression through their ability to bind to acetylated chromatin and subsequently activate RNA PolII-driven transcriptional elongation. Small molecule BET inhibitors prevent binding of BET proteins to acetylated histones and inhibit transcriptional activation of BET target genes. BET inhibitors attenuate cell growth and survival in several hematologic cancer models, partially through the down-regulation of the critical oncogene, MYC. We hypothesized that BET inhibitors will regulate MYC expression in solid tumors that frequently over-express MYC. Here we describe the effects of the highly specific BET inhibitor, I-BET762, on MYC expression in prostate cancer models. I-BET762 potently reduced MYC expression in prostate cancer cell lines and a patient-derived tumor model with subsequent inhibition of cell growth and reduction of tumor burden in vivo. Our data suggests that I-BET762 effects are partially driven by MYC down-regulation and underlines the critical importance of additional mechanisms of I-BET762 induced phenotypes.


Subject(s)
Benzodiazepines/pharmacology , Nuclear Proteins/antagonists & inhibitors , Prostatic Neoplasms, Castration-Resistant/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Down-Regulation , Gene Expression Profiling , Humans , Male , Mice , Mice, SCID , Prostatic Neoplasms, Castration-Resistant/enzymology , Prostatic Neoplasms, Castration-Resistant/pathology , Xenograft Model Antitumor Assays
14.
PLoS One ; 8(8): e72967, 2013.
Article in English | MEDLINE | ID: mdl-24009722

ABSTRACT

BET family proteins are epigenetic regulators known to control expression of genes involved in cell growth and oncogenesis. Selective inhibitors of BET proteins exhibit potent anti-proliferative activity in a number of hematologic cancer models, in part through suppression of the MYC oncogene and downstream Myc-driven pathways. However, little is currently known about the activity of BET inhibitors in solid tumor models, and whether down-regulation of MYC family genes contributes to sensitivity. Here we provide evidence for potent BET inhibitor activity in neuroblastoma, a pediatric solid tumor associated with a high frequency of MYCN amplifications. We treated a panel of neuroblastoma cell lines with a novel small molecule inhibitor of BET proteins, GSK1324726A (I-BET726), and observed potent growth inhibition and cytotoxicity in most cell lines irrespective of MYCN copy number or expression level. Gene expression analyses in neuroblastoma cell lines suggest a role of BET inhibition in apoptosis, signaling, and N-Myc-driven pathways, including the direct suppression of BCL2 and MYCN. Reversal of MYCN or BCL2 suppression reduces the potency of I-BET726-induced cytotoxicity in a cell line-specific manner; however, neither factor fully accounts for I-BET726 sensitivity. Oral administration of I-BET726 to mouse xenograft models of human neuroblastoma results in tumor growth inhibition and down-regulation MYCN and BCL2 expression, suggesting a potential role for these genes in tumor growth. Taken together, our data highlight the potential of BET inhibitors as novel therapeutics for neuroblastoma, and suggest that sensitivity is driven by pleiotropic effects on cell growth and apoptotic pathways in a context-specific manner.


Subject(s)
Benzodiazepines/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Neuroblastoma/genetics , Neuroblastoma/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , RNA-Binding Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Apoptosis/genetics , Benzodiazepines/chemistry , Benzodiazepines/toxicity , Cell Cycle Proteins , Cell Proliferation/drug effects , Cluster Analysis , Disease Models, Animal , Female , Gene Expression Profiling , Gene Regulatory Networks , Humans , Kinetics , Mice , Models, Molecular , Molecular Conformation , N-Myc Proto-Oncogene Protein , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/chemistry , Transcription Factors/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
15.
Mol Endocrinol ; 24(8): 1665-74, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20610535

ABSTRACT

Androgen signaling through the androgen receptor (AR), a ligand-dependent transcription factor within the steroid receptor superfamily, plays an important role in the development and maintenance of many tissues. In muscle, androgens act as anabolic agents that increase both muscle mass and strength; however, a key unanswered question is the mechanism through which AR-mediated gene expression leads to these effects. To gain further insight into the mechanism of AR action in muscle, we identified AR-binding sites in primary human muscle cells using ChIP-on-Chip (chromatin immunoprecipitation coupled with tiling microarray detection of genomic fragments). Through this analysis, we identified 32,518 potential AR-binding sites throughout the genome that were enriched upon androgen treatment. Sequence analysis of these regions indicated that approximately 90% possess a consensus androgen response element or half-site. Among the identified AR-binding sites are genes known to be directly regulated by AR, confirming the validity of our methodology. Additionally, we identified a number of novel AR targets, including genes and micro-RNAs implicated in muscle differentiation and function, suggesting a direct role for AR-mediated transcription in muscle development. Intriguingly, binding sequences for the Mef2 family of transcription factors were enriched in the AR-bound regions, and we show that several Mef2c-dependent genes are direct targets of AR, suggesting a functional interaction between Mef2c and AR in skeletal muscle. Our results provide new insights into the mechanisms by which androgens promote muscle growth and validate AR as a potential therapeutic target for sarcopenia, muscle wasting, and other androgen-related muscle disorders.


Subject(s)
Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Receptors, Androgen/metabolism , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Humans , MADS Domain Proteins/genetics , MADS Domain Proteins/metabolism , MEF2 Transcription Factors , Myoblasts/cytology , Myoblasts/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Polymerase Chain Reaction , Protein Binding , Receptors, Androgen/genetics
16.
Mol Cell ; 29(1): 102-11, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-18206973

ABSTRACT

Polycomb genes encode critical regulators of both normal stem cells and cancer stem cells. A gene signature that includes Polycomb genes and additional genes coregulated with Polycomb genes was recently identified. The expression of this signature has been reported to identify tumors with the cancer stem cell phenotypes of aggressive growth, metastasis, and therapy resistance. Most members of this 11 gene signature encode proteins with well-defined roles in human cancer. However, the function of the signature member USP22 remains unknown. We report that USP22 is a previously uncharacterized subunit of the human SAGA transcriptional cofactor complex. Within SAGA, USP22 deubiquitylates histone H2B. Furthermore, USP22 is recruited to specific genes by activators such as the Myc oncoprotein, where it is required for transcription. In support of a functional role within the Polycomb/cancer stem cell signature, USP22 is required for appropriate progression through the cell cycle.


Subject(s)
Cell Cycle/physiology , Gene Expression Profiling , Neoplastic Stem Cells/metabolism , Protein Processing, Post-Translational/physiology , Thiolester Hydrolases/physiology , Trans-Activators/physiology , Transcription Factors/chemistry , Transcription, Genetic/physiology , Ubiquitination/physiology , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/physiology , Biomarkers , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Transformed/cytology , Cell Line, Transformed/metabolism , Cell Line, Tumor/cytology , Cell Line, Tumor/metabolism , Histone Acetyltransferases/analysis , Histone Acetyltransferases/physiology , Humans , Lung Neoplasms/pathology , Neoplastic Stem Cells/cytology , Nuclear Proteins/analysis , Nuclear Proteins/physiology , Promoter Regions, Genetic/genetics , Protein Subunits , Proto-Oncogene Proteins c-myc/physiology , Recombinant Fusion Proteins/physiology , Transcription Factors/physiology , Ubiquitin Thiolesterase , p300-CBP Transcription Factors/analysis , p300-CBP Transcription Factors/physiology
17.
Mol Cell ; 27(2): 275-288, 2007 Jul 20.
Article in English | MEDLINE | ID: mdl-17643376

ABSTRACT

Histone modifications play an important role in transcription. We previously studied histone H2B ubiquitylation on lysine 123 and subsequent deubiquitylation by SAGA-associated Ubp8. Unlike other histone modifications, both the addition and removal of ubiquitin are required for optimal transcription. Here we report that deubiquitylation of H2B is important for recruitment of a complex containing the kinase Ctk1, resulting in phosphorylation of the RNA polymerase II (Pol II) C-terminal domain (CTD), and for subsequent recruitment of the Set2 methyltransferase. We find that Ctk1 interacts with histones H2A and H2B, and that persistent H2B ubiquitylation disrupts these interactions. We further show that Ubp8 enters the GAL1 coding region through an interaction with Pol II. These findings reveal a mechanism by which H2B ubiquitylation acts as a barrier to Ctk1 association with active genes, while subsequent deubiquitylation by Ubp8 triggers Ctk1 recruitment at the appropriate point in activation.


Subject(s)
Endopeptidases/metabolism , Histones/metabolism , Protein Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/metabolism , Ubiquitin/metabolism , Endopeptidases/chemistry , Endopeptidases/genetics , Genes, Fungal , Histones/chemistry , Histones/genetics , Models, Biological , Multiprotein Complexes , Nucleosomes/metabolism , Open Reading Frames , Protein Kinases/chemistry , Protein Kinases/genetics , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription, Genetic
18.
Mol Cell ; 17(4): 585-94, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15721261

ABSTRACT

Low levels of histone covalent modifications are associated with gene silencing at telomeres and other regions in the yeast S. cerevisiae. Although the histone deacetylase Sir2 maintains low acetylation, mechanisms responsible for low H2B ubiquitylation and low H3 methylation are unknown. Here, we show that the ubiquitin protease Ubp10 targets H2B for deubiquitylation, helping to localize Sir2 to the telomere. Ubp10 exhibits reciprocal Sir2-dependent preferential localization proximal to telomeres, where Ubp10 serves to maintain low H2B Lys123 ubiquitylation in this region and, through previously characterized crosstalk, maintains low H3 Lys4 and Lys79 methylation in a slightly broader region. Ubp10 is also localized to the rDNA locus, a second silenced domain, where it similarly maintains low histone methylation. We compare Ubp10 to Ubp8, the SAGA-associated H2B deubiquitylase involved in gene activation, and show that telomeric and gene-silencing functions are specific to Ubp10. Our results suggest that these H2B-deubiquitylating enzymes have distinct genomic functions.


Subject(s)
Gene Expression Regulation, Fungal/physiology , Gene Silencing , Histone Deacetylases/physiology , Histones/metabolism , Nuclear Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Silent Information Regulator Proteins, Saccharomyces cerevisiae/physiology , Sirtuins/physiology , Telomere/physiology , Ubiquitin/metabolism , Acetylation , Chromatin Immunoprecipitation , DNA, Ribosomal , Down-Regulation , Lysine/metabolism , Methylation , Proteasome Endopeptidase Complex , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sirtuin 2 , Transcription, Genetic , Transcriptional Activation , Ubiquitin Thiolesterase
19.
J Biol Chem ; 277(10): 8243-7, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11756452

ABSTRACT

We have recently described a large (20 S) protein arginine methyltransferase complex, termed the methylosome, that contains the methyltransferase JBP1 (PRMT5) and the pICln protein. The methylosome functions to modify specific arginines to dimethylarginines in the arginine- and glycine-rich domains of several spliceosomal Sm proteins, and this modification targets these proteins to the survival of motor neurons (SMN) complex for assembly into small nuclear ribonucleoprotein (snRNP) core particles. Here, we describe a novel component of the methylosome, a 50-kilodalton WD repeat protein termed methylosome protein 50 (MEP50). We show that MEP50 is important for methylosome activity and binds to JBP1 and to a subset of Sm proteins. Because WD repeat proteins provide a platform for multiple protein interactions, MEP50 may function to mediate the interaction of multiple substrates with the methylosome. Interestingly, all of the known components of the methylosome bind Sm proteins, suggesting that in addition to producing properly methylated substrates for the SMN complex, the methylosome may be involved in Sm protein rearrangements or pre-assembly required for snRNP biogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/chemistry , Protein-Arginine N-Methyltransferases/chemistry , Ribonucleoproteins, Small Nuclear/metabolism , Amino Acid Sequence , Arginine/chemistry , Carrier Proteins/metabolism , Centrifugation, Density Gradient , DNA/metabolism , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/metabolism , HeLa Cells , Humans , Immunoblotting , Methylation , Molecular Sequence Data , Plasmids/metabolism , Protein Binding , Protein Methyltransferases/metabolism , Protein Structure, Tertiary , Repetitive Sequences, Amino Acid , Ribonucleoproteins, Small Nuclear/chemistry
20.
Genes Dev ; 17(21): 2648-63, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14563679

ABSTRACT

Gene activation and repression regulated by acetylation and deacetylation represent a paradigm for the function of histone modifications. We provide evidence that, in contrast, histone H2B monoubiquitylation and its deubiquitylation are both involved in gene activation. Substitution of the H2B ubiquitylation site at Lys 123 (K123) lowered transcription of certain genes regulated by the acetylation complex SAGA. Gene-associated H2B ubiquitylation was transient, increasing early during activation, and then decreasing coincident with significant RNA accumulation. We show that Ubp8, a component of the SAGA acetylation complex, is required for SAGA-mediated deubiquitylation of histone H2B in vitro. Loss of Ubp8 in vivo increased both gene-associated and overall cellular levels of ubiquitylated H2B. Deletion of Ubp8 lowered transcription of SAGA-regulated genes, and the severity of this defect was exacerbated by codeletion of the Gcn5 acetyltransferase within SAGA. In addition, disruption of either ubiquitylation or Ubp8-mediated deubiquitylation of H2B resulted in altered levels of gene-associated H3 Lys 4 methylation and Lys 36 methylation, which have both been linked to transcription. These results suggest that the histone H2B ubiquitylation state is dynamic during transcription, and that the sequence of histone modifications helps to control transcription.


Subject(s)
Gene Expression Regulation , Histones/metabolism , Ubiquitin/metabolism , Lysine/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcription, Genetic , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL